Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Malar J ; 15(1): 377, 2016 Jul 22.
Article in English | MEDLINE | ID: mdl-27448805

ABSTRACT

BACKGROUND: In this phase 1 clinical trial, healthy adult, malaria-naïve subjects were immunized with radiation-attenuated Plasmodium falciparum sporozoites (PfRAS) by mosquito bite and then underwent controlled human malaria infection (CHMI). The PfRAS model for immunization against malaria had previously induced >90 % sterile protection against homologous CHMI. This study was to further explore the safety, tolerability and protective efficacy of the PfRAS model and to provide biological specimens to characterize protective immune responses and identify protective antigens in support of malaria vaccine development. METHODS: Fifty-seven subjects were screened, 41 enrolled and 30 received at least one immunization. The true-immunized subjects received PfRAS via mosquito bite and the mock-immunized subjects received mosquito bites from irradiated uninfected mosquitoes. Sera and peripheral blood mononuclear cells (PBMCs) were collected before and after PfRAS immunizations. RESULTS: Immunization with PfRAS was generally safe and well tolerated, and repeated immunization via mosquito bite did not appear to increase the risk or severity of AEs. Local adverse events (AEs) of true-immunized and mock-immunized groups consisted of erythaema, papules, swelling, and induration and were consistent with reactions from mosquito bites seen in nature. Two subjects, one true- and one mock-immunized, developed large local reactions that completely resolved, were likely a result of mosquito salivary antigens, and were withdrawn from further participation as a safety precaution. Systemic AEs were generally rare and mild, consisting of headache, myalgia, nausea, and low-grade fevers. Two true-immunized subjects experienced fever, malaise, myalgia, nausea, and rigours approximately 16 h after immunization. These symptoms likely resulted from pre-formed antibodies interacting with mosquito salivary antigens. Ten subjects immunized with PfRAS underwent CHMI and five subjects (50 %) were sterilely protected and there was a significant delay to parasitaemia in the other five subjects. All ten subjects developed humoral immune responses to whole sporozoites and to the circumsporozoite protein prior to CHMI, although the differences between protected and non-protected subjects were not statistically significant for this small sample size. CONCLUSIONS: The protective efficacy of this clinical trial (50 %) was notably less than previously reported (>90 %). This may be related to differences in host genetics or the inherent variability in mosquito biting behavior and numbers of sporozoites injected. Differences in trial procedures, such as the use of leukapheresis prior to CHMI and of a longer interval between the final immunization and CHMI in these subjects compared to earlier trials, may also have reduced protective efficacy. This trial has been retrospectively registered at ISRCTN ID 17372582, May 31, 2016.


Subject(s)
Antibodies, Protozoan/blood , Culicidae/physiology , Insect Bites and Stings , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Adolescent , Adult , Animals , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Humans , Malaria Vaccines/administration & dosage , Male , Middle Aged , Plasmodium falciparum/radiation effects , Sporozoites/immunology , Sporozoites/radiation effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Young Adult
2.
PLoS One ; 10(8): e0136109, 2015.
Article in English | MEDLINE | ID: mdl-26292257

ABSTRACT

BACKGROUND: Nearly 100% protection against malaria infection can be achieved in humans by immunization with P. falciparum radiation-attenuated sporozoites (RAS). Although it is thought that protection is mediated by T cell and antibody responses, only a few of the many pre-erythrocytic (sporozoite and liver stage) antigens that are targeted by these responses have been identified. METHODOLOGY: Twenty seven P. falciparum pre-erythrocytic antigens were selected using bioinformatics analysis and expression databases and were expressed in a wheat germ cell-free protein expression system. Recombinant proteins were recognized by plasma from RAS-immunized subjects, and 21 induced detectable antibody responses in mice and rabbit and sera from these immunized animals were used to characterize these antigens. All 21 proteins localized to the sporozoite: five localized to the surface, seven localized to the micronemes, cytoplasm, endoplasmic reticulum or nucleus, two localized to the surface and cytoplasm, and seven remain undetermined. PBMC from RAS-immunized volunteers elicited positive ex vivo or cultured ELISpot responses against peptides from 20 of the 21 antigens. CONCLUSIONS: These T cell and antibody responses support our approach of using reagents from RAS-immunized subjects to screen potential vaccine antigens, and have led to the identification of a panel of novel P. falciparum antigens. These results provide evidence to further evaluate these antigens as vaccine candidates. TRIAL REGISTRATION: ClinicalTrials.gov NCT00870987 ClinicalTrials.gov NCT00392015.


Subject(s)
Antigens, Protozoan/immunology , Erythrocytes/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Animals , Erythrocytes/parasitology , Humans , Immunization , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/parasitology , Malaria Vaccines/pharmacology , Malaria, Falciparum/blood , Mice , Mice, Inbred BALB C , Protozoan Proteins/immunology , Rabbits , Sporozoites/immunology , T-Lymphocytes/immunology , T-Lymphocytes/parasitology
3.
PLoS One ; 8(2): e55571, 2013.
Article in English | MEDLINE | ID: mdl-23457473

ABSTRACT

BACKGROUND: Gene-based vaccination using prime/boost regimens protects animals and humans against malaria, inducing cell-mediated responses that in animal models target liver stage malaria parasites. We tested a DNA prime/adenovirus boost malaria vaccine in a Phase 1 clinical trial with controlled human malaria infection. METHODOLOGY/PRINCIPAL FINDINGS: The vaccine regimen was three monthly doses of two DNA plasmids (DNA) followed four months later by a single boost with two non-replicating human serotype 5 adenovirus vectors (Ad). The constructs encoded genes expressing P. falciparum circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1). The regimen was safe and well-tolerated, with mostly mild adverse events that occurred at the site of injection. Only one AE (diarrhea), possibly related to immunization, was severe (Grade 3), preventing daily activities. Four weeks after the Ad boost, 15 study subjects were challenged with P. falciparum sporozoites by mosquito bite, and four (27%) were sterilely protected. Antibody responses by ELISA rose after Ad boost but were low (CSP geometric mean titer 210, range 44-817; AMA1 geometric mean micrograms/milliliter 11.9, range 1.5-102) and were not associated with protection. Ex vivo IFN-γ ELISpot responses after Ad boost were modest (CSP geometric mean spot forming cells/million peripheral blood mononuclear cells 86, range 13-408; AMA1 348, range 88-1270) and were highest in three protected subjects. ELISpot responses to AMA1 were significantly associated with protection (p = 0.019). Flow cytometry identified predominant IFN-γ mono-secreting CD8+ T cell responses in three protected subjects. No subjects with high pre-existing anti-Ad5 neutralizing antibodies were protected but the association was not statistically significant. SIGNIFICANCE: The DNA/Ad regimen provided the highest sterile immunity achieved against malaria following immunization with a gene-based subunit vaccine (27%). Protection was associated with cell-mediated immunity to AMA1, with CSP probably contributing. Substituting a low seroprevalence vector for Ad5 and supplementing CSP/AMA1 with additional antigens may improve protection. TRIAL REGISTRATION: ClinicalTrials.govNCT00870987.


Subject(s)
Adenoviruses, Human/genetics , Antigens, Protozoan/genetics , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Membrane Proteins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Vaccines, DNA/therapeutic use , Adenoviruses, Human/immunology , Adolescent , Adult , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Immunity, Cellular , Interferon-gamma/immunology , Malaria Vaccines/adverse effects , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Membrane Proteins/immunology , Middle Aged , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Vaccines, DNA/adverse effects , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Young Adult
4.
PLoS One ; 6(10): e24586, 2011.
Article in English | MEDLINE | ID: mdl-22003383

ABSTRACT

BACKGROUND: Models of immunity to malaria indicate the importance of CD8+ T cell responses for targeting intrahepatic stages and antibodies for targeting sporozoite and blood stages. We designed a multistage adenovirus 5 (Ad5)-vectored Plasmodium falciparum malaria vaccine, aiming to induce both types of responses in humans, that was tested for safety and immunogenicity in a Phase 1 dose escalation trial in Ad5-seronegative volunteers. METHODOLOGY/PRINCIPAL FINDINGS: The NMRC-M3V-Ad-PfCA vaccine combines two adenovectors encoding circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1). Group 1 (n = 6) healthy volunteers received one intramuscular injection of 2×10∧10 particle units (1×10∧10 each construct) and Group 2 (n = 6) a five-fold higher dose. Transient, mild to moderate adverse events were more pronounced with the higher dose. ELISpot responses to CSP and AMA1 peaked at 1 month, were higher in the low dose (geomean CSP = 422, AMA1 = 862 spot forming cells/million) than in the high dose (CSP = 154, p = 0.049, AMA1 = 423, p = 0.045) group and were still positive at 12 months in a number of volunteers. ELISpot depletion assays identified dependence on CD4+ or on both CD4+ and CD8+ T cells, with few responses dependent only on CD8+ T cells. Intracellular cytokine staining detected stronger CD8+ than CD4+ T cell IFN-γ responses (CSP p = 0.0001, AMA1 p = 0.003), but similar frequencies of multifunctional CD4+ and CD8+ T cells secreting two or more of IFN-γ, TNF-α or IL-2. Median fluorescence intensities were 7-10 fold higher in triple than single secreting cells. Antibody responses were low but trended higher in the high dose group and did not inhibit growth of cultured P. falciparum blood stage parasites. SIGNIFICANCE: As found in other trials, adenovectored vaccines appeared safe and well-tolerated at doses up to 1×10∧11 particle units. This is the first demonstration in humans of a malaria vaccine eliciting strong CD8+ T cell IFN-γ responses. TRIAL REGISTRATION: ClinicalTrials.govNCT00392015.


Subject(s)
Adenoviridae/genetics , Antigens, Protozoan/adverse effects , Antigens, Protozoan/immunology , Genetic Vectors/genetics , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Adolescent , Adult , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dose-Response Relationship, Immunologic , Female , Gene Expression , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Interferon-gamma/metabolism , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Male , Membrane Proteins/adverse effects , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/immunology , Middle Aged , Peptide Fragments/immunology , Protozoan Proteins/adverse effects , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Young Adult
5.
PLoS One ; 6(10): e25868, 2011.
Article in English | MEDLINE | ID: mdl-22003411

ABSTRACT

BACKGROUND: A protective malaria vaccine will likely need to elicit both cell-mediated and antibody responses. As adenovirus vaccine vectors induce both these responses in humans, a Phase 1/2a clinical trial was conducted to evaluate the efficacy of an adenovirus serotype 5-vectored malaria vaccine against sporozoite challenge. METHODOLOGY/PRINCIPAL FINDINGS: NMRC-MV-Ad-PfC is an adenovirus vector encoding the Plasmodium falciparum 3D7 circumsporozoite protein (CSP). It is one component of a two-component vaccine NMRC-M3V-Ad-PfCA consisting of one adenovector encoding CSP and one encoding apical membrane antigen-1 (AMA1) that was evaluated for safety and immunogenicity in an earlier study (see companion paper, Sedegah et al). Fourteen Ad5 seropositive or negative adults received two doses of NMRC-MV-Ad-PfC sixteen weeks apart, at 1 x 1010 particle units per dose. The vaccine was safe and well tolerated. All volunteers developed positive ELISpot responses by 28 days after the first immunization (geometric mean 272 spot forming cells/million[sfc/m]) that declined during the following 16 weeks and increased after the second dose to levels that in most cases were less than the initial peak (geometric mean 119 sfc/m). CD8+ predominated over CD4+ responses, as in the first clinical trial. Antibody responses were poor and like ELISpot responses increased after the second immunization but did not exceed the initial peak. Pre-existing neutralizing antibodies (NAb) to Ad5 did not affect the immunogenicity of the first dose, but the fold increase in NAb induced by the first dose was significantly associated with poorer antibody responses after the second dose, while ELISpot responses remained unaffected. When challenged by the bite of P. falciparum-infected mosquitoes, two of 11 volunteers showed a delay in the time to patency compared to infectivity controls, but no volunteers were sterilely protected. SIGNIFICANCE: The NMRC-MV-Ad-PfC vaccine expressing CSP was safe and well tolerated given as two doses, but did not provide sterile protection. TRIAL REGISTRATION: ClinicalTrials.gov NCT00392015.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/adverse effects , Protozoan Proteins/immunology , Adolescent , Adult , Antigens, Protozoan/adverse effects , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Dose-Response Relationship, Immunologic , Female , Gene Expression , Humans , Malaria Vaccines/genetics , Male , Membrane Proteins/adverse effects , Membrane Proteins/genetics , Membrane Proteins/immunology , Middle Aged , Plasmodium falciparum/cytology , Protozoan Proteins/genetics , Sporozoites/immunology , Young Adult
7.
Mol Biochem Parasitol ; 158(1): 32-45, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18164079

ABSTRACT

We have evaluated a technology called transcriptionally active PCR (TAP) for high throughput identification and prioritization of novel target antigens from genomic sequence data using the Plasmodium parasite, the causative agent of malaria, as a model. First, we adapted the TAP technology for the highly AT-rich Plasmodium genome, using well-characterized P. falciparum and P. yoelii antigens and a small panel of uncharacterized open reading frames from the P. falciparum genome sequence database. We demonstrated that TAP fragments encoding six well-characterized P. falciparum antigens and five well-characterized P. yoelii antigens could be amplified in an equivalent manner from both plasmid DNA and genomic DNA templates, and that uncharacterized open reading frames could also be amplified from genomic DNA template. Second, we showed that the in vitro expression of the TAP fragments was equivalent or superior to that of supercoiled plasmid DNA encoding the same antigen. Third, we evaluated the in vivo immunogenicity of TAP fragments encoding a subset of the model P. falciparum and P. yoelii antigens. We found that antigen-specific antibody and cellular immune responses induced by the TAP fragments in mice were equivalent or superior to those induced by the corresponding plasmid DNA vaccines. Finally, we developed and demonstrated proof-of-principle for an in vitro humoral immunoscreening assay for down-selection of novel target antigens. These data support the potential of a TAP approach for rapid high throughput functional screening and identification of potential candidate vaccine antigens from genomic sequence data.


Subject(s)
Antigens, Protozoan/genetics , Malaria Vaccines/immunology , Plasmodium falciparum/genetics , Plasmodium yoelii/genetics , Polymerase Chain Reaction/methods , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Cytokines/biosynthesis , Female , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium falciparum/immunology , Plasmodium yoelii/immunology , Spleen/immunology , T-Lymphocytes/immunology , Vaccines, DNA/genetics
8.
Microbes Infect ; 9(12-13): 1439-46, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17913540

ABSTRACT

Effective vaccines against infectious diseases and biological warfare agents remain an urgent public health priority. Studies have characterized the differentiation of effector and memory T cells and identified a subset of T cells capable of conferring enhanced protective immunity against pathogen challenge. We hypothesized that the kinetics of T cell differentiation influences the immunogenicity and protective efficacy of plasmid DNA vaccines, and tested this hypothesis in the Plasmodium yoelii murine model of malaria. We found that increasing the interval between immunizations significantly enhanced the frequency and magnitude of CD8+ and CD4+ T cell responses as well as protective immunity against sporozoite challenge. Moreover, the interval between immunizations was more important than the total number of immunizations. Immunization interval had a significantly greater impact on T cell responses and protective immunity than on antibody responses. With prolonged immunization intervals, T cell responses induced by homologous DNA only regimens achieved levels similar to those induced by heterologous DNA prime/ virus boost immunization at standard intervals. Our studies establish that the dosing interval significantly impacts the immunogenicity and protective efficacy of plasmid DNA vaccines.


Subject(s)
Immunization Schedule , Malaria/immunology , Malaria/prevention & control , Plasmids/immunology , Plasmodium yoelii/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Immunization , Immunization, Secondary , Malaria/parasitology , Malaria Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium yoelii/pathogenicity
9.
Malar J ; 6: 135, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17925026

ABSTRACT

The present study has evaluated the immunogenicity of single or multiple Plasmodium falciparum (Pf) antigens administered in a DNA prime/poxvirus boost regimen with or without the poloxamer CRL1005 in rhesus monkeys. Animals were primed with PfCSP plasmid DNA or a mixture of PfCSP, PfSSP2/TRAP, PfLSA1, PfAMA1 and PfMSP1-42 (CSLAM) DNA vaccines in PBS or formulated with CRL1005, and subsequently boosted with ALVAC-Pf7, a canarypox virus expressing the CSLAM antigens. Cell-mediated immune responses were evaluated by IFN-gamma ELIspot and intracellular cytokine staining, using recombinant proteins and overlapping synthetic peptides. Antigen-specific and parasite-specific antibody responses were evaluated by ELISA and IFAT, respectively. Immune responses to all components of the multi-antigen mixture were demonstrated following immunization with either DNA/PBS or DNA/CRL1005, and no antigen interference was observed in animals receiving CSLAM as compared to PfCSP alone. These data support the down-selection of the CSLAM antigen combination. CRL1005 formulation had no apparent effect on vaccine-induced T cell or antibody responses, either before or after viral boost. In high responder monkeys, CD4+IL-2+ responses were more predominant than CD8+ T cell responses. Furthermore, CD8+ IFN-gamma responses were detected only in the presence of detectable CD4+ T cell responses. Overall, this study demonstrates the potential for multivalent Pf vaccines based on rational antigen selection and combination, and suggests that further formulation development to increase the immunogenicity of DNA encoded antigens is warranted.


Subject(s)
Antigens, Protozoan/immunology , Immunization, Secondary/methods , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Poxviridae/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/genetics , Immunization , Macaca mulatta , Malaria Vaccines/immunology , Plasmids , Vaccines, DNA/administration & dosage
10.
Acad Radiol ; 14(3): 330-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17307666

ABSTRACT

The proceedings of a workshop focusing on a project to evaluate the use of fluorodeoxyglucose-positron emission tomography (FDG-PET) as a tool to measure treatment response in non-Hodgkin lymphoma (NHL) are described. Sponsored by the Leukemia & Lymphoma Society, the Foundation of the National Institutes of Health, and the National Cancer Institute, and attended by representatives of the Food and Drug Administration, the Centers for Medicare and Medicaid Services, and scientists and clinical researchers from academia and the pharmaceutical and medical imaging industries, the workshop reviewed the etiology and current standards of care for NHL and proposed the development of a clinical trial to validate FDG-PET imaging techniques as a predictive biomarker for cancer therapy response. As organized under the auspices of the Oncology Biomarker Qualification Initiative, the three federal health agencies and their private sector and nonprofit/advocacy group partners believe that FDG-PET not only demonstrates the potential to be used for the diagnosis and staging of many cancers but in particular can provide an early indication of therapeutic response that is well correlated with clinical outcomes for chemotherapy for this common form of lymphoma. The development of standardized criteria for FDG-PET imaging and establishment of procedures for transmission, storage, quality assurance, and analysis of PET images afforded by this demonstration project could streamline clinical trials of new treatments for more intractable forms of lymphoma and other cancers and, hence, accelerate new drug approvals.


Subject(s)
Fluorodeoxyglucose F18 , Lymphoma, Non-Hodgkin/diagnostic imaging , Positron-Emission Tomography , Clinical Trials, Phase II as Topic , Female , Humans , Lymphoma, Non-Hodgkin/drug therapy , Male , Quality Assurance, Health Care , Reproducibility of Results
11.
Acta Trop ; 100(1-2): 70-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17113021

ABSTRACT

Antibody levels against malaria antigens were measured among patients presenting with uncomplicated malaria at health centers from three locations in Zimbabwe (Bindura, Chiredzi and Kariba) that are distinct with regard to altitude and climatic conditions. Antibody levels were determined by ELISA using the antigens, apical membrane antigen 1 (AMA-1), erythrocyte binding antigen 175 (EBA-175), circumsporozoite surface protein (CSP), merozoite surface protein 1 (MSP-1) and Pfg27. For all the antigens tested, IgG and IgM levels were higher for Bindura (altitude 1100 m) compared to Kariba (<600 m, altitude) and Chiredzi (approximately 600 m, altitude) with the exception of IgG and IgM to AMA-1 and EBA-175 which were similar between Chiredzi and Bindura. Plasma samples were further analyzed for their functional activity by testing their ability to inhibit the growth of Plasmodium falciparum in culture. Our results, determined by microscopy and verified by the LDH assay revealed that plasma from the three locations had similar inhibitory activity against the growth of P. falciparum in vitro. Our data revealed that highest growth inhibition correlated with the highest levels of MSP-1 antibody values.


Subject(s)
Altitude , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Adolescent , Adult , Animals , Antigens, Protozoan/administration & dosage , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Incidence , Malaria Vaccines/administration & dosage , Malaria, Falciparum/epidemiology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Male , Zimbabwe/epidemiology
12.
Infect Immun ; 74(11): 6331-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16966412

ABSTRACT

Two different cell populations respond to potent T-cell-inducing vaccinations. The induction and loss of effector cells can be seen using an ex vivo enzyme-linked immunospot (ELISPOT) assay, but the more durable resting memory response is demonstrable by a cultured ELISPOT assay. The relationship of the early effector response to durable resting memory is incompletely understood. Effector phenotype is usually identified by gamma interferon (IFN-gamma) production, but interleukin-2 (IL-2) has been specifically linked to the differentiation of memory cells. Here, IFN-gamma- and IL-2-secreting effector cells were identified by an ex vivo ELISPOT assay 1 week after vaccination and compared with the resting memory responses detected by a cultured ELISPOT assay 3 months later. The different kinetics and induction of IL-2 by different vaccines and natural exposure are described. Furthermore, both early IFN-gamma and IL-2 production independently predicted subsequent memory responses at 3 months in malaria-naïve volunteers, but only IFN-gamma predicted memory in malaria-exposed volunteers. However, dual ELISPOT assays were also performed on malaria-exposed volunteers to identify cells producing both cytokines simultaneously. This demonstrated that double-cytokine-producing cells were highly predictive of memory. This assay may be useful in predicting vaccinations most likely to generate stable, long-term memory responses.


Subject(s)
Immunologic Memory , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria/immunology , Animals , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Malaria/metabolism , Malaria/prevention & control , Plasmodium/immunology , Predictive Value of Tests , Resting Phase, Cell Cycle/immunology , Time Factors
13.
Vaccine ; 24(11): 1921-7, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16298024

ABSTRACT

We evaluated the capacity of the cationic lipid based formulation, Vaxfectin, to enhance the immunogenicity and protective efficacy of DNA-based vaccine regimens in the Plasmodium yoelii murine malaria model. We immunized Balb/c mice with varying doses (0.4-50 microg) of plasmid DNA (pDNA) encoding the P. yoelii circumsporozoite protein (PyCSP), either in a homologous DNA/DNA regimen (D-D) or a heterologous prime-boost DNA-poxvirus regimen (D-V). At the lowest pDNA doses, Vaxfectin substantially enhanced IFA titers, ELISPOT frequencies, and protective efficacy. Clinical trials of pDNA vaccines have often used low pDNA doses based on a per kilogram weight basis. Formulation of pDNA vaccines in Vaxfectin may improve their potency in human clinical trials.


Subject(s)
Malaria Vaccines/immunology , Malaria/prevention & control , Phosphatidylethanolamines/pharmacology , Plasmodium yoelii/immunology , Protozoan Proteins/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Female , Fluorescent Antibody Technique, Indirect , Humans , Immunization, Secondary , Lymphocytes/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Phosphatidylethanolamines/administration & dosage , Protozoan Proteins/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccinia virus/genetics
14.
J Biol Chem ; 280(48): 40169-76, 2005 Dec 02.
Article in English | MEDLINE | ID: mdl-16203726

ABSTRACT

Glycosylphosphatidylinositol (GPI)-anchored proteins coat the surface of extracellular Plasmodium falciparum merozoites, of which several are highly validated candidates for inclusion in a blood-stage malaria vaccine. Here we determined the proteome of gradient-purified detergent-resistant membranes of mature blood-stage parasites and found that these membranes are greatly enriched in GPI-anchored proteins and their putative interacting partners. Also prominent in detergent-resistant membranes are apical organelle (rhoptry), multimembrane-spanning, and proteins destined for export into the host erythrocyte cytosol. Four new GPI-anchored proteins were identified, and a number of other novel proteins that are predicted to localize to the merozoite surface and/or apical organelles were detected. Three of the putative surface proteins possessed six-cysteine (Cys6) motifs, a distinct fold found in adhesive surface proteins expressed in other life stages. All three Cys6 proteins, termed Pf12, Pf38, and Pf41, were validated as merozoite surface antigens recognized strongly by antibodies present in naturally infected individuals. In addition to the merozoite surface, Pf38 was particularly prominent in the secretory apical organelles. A different cysteine-rich putative GPI-anchored protein, Pf92, was also localized to the merozoite surface. This insight into merozoite surfaces provides new opportunities for understanding both erythrocyte invasion and anti-parasite immunity.


Subject(s)
Antigens, Protozoan/chemistry , Merozoite Surface Protein 1/chemistry , Plasmodium falciparum/metabolism , Amino Acid Motifs , Animals , Antigens, Protozoan/metabolism , Antigens, Surface/chemistry , Cell Membrane/metabolism , Cysteine/chemistry , Detergents/pharmacology , Epidermal Growth Factor/chemistry , Erythrocytes/metabolism , Glycosylphosphatidylinositols/chemistry , Green Fluorescent Proteins/chemistry , Membrane Microdomains/chemistry , Models, Biological , Protein Binding , Protein Structure, Tertiary , Proteins , Proteomics , Protozoan Proteins/chemistry
15.
Acta Trop ; 95(3): 260-4, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16018960

ABSTRACT

The promise of new interventions against malaria and other infectious diseases of global health importance derived from pathogen genomic sequence data may only be realized through the coordinated effort of genomic and post-genomics scientists, vaccine and drug developers along with lab- and field-based investigators. With the availability of the Plasmodium falciparum genome and the genomes of related species, post-genomics research can now be applied to the development of new interventions against malaria and may provide a more complete understanding of complex parasite biology. The vast amount of data that are generated through these new approaches must be organized, assembled and made accessible in a useful manner. By establishing a set of "credentials" for each gene in the genome, which captures information about gene expression, diversity, function and other information, the time and resources needed to test and evaluate candidate drugs and vaccines can be substantially reduced and a more complete picture of parasite biology can be created.


Subject(s)
Genomics/trends , Malaria Vaccines , Malaria/genetics , Plasmodium falciparum/immunology , Animals , Genetic Variation , Humans , Malaria/prevention & control , Plasmodium falciparum/genetics
16.
Mol Biochem Parasitol ; 143(1): 67-79, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16005087

ABSTRACT

The sexual stages of malarial parasites are essential for the mosquito transmission of the disease and therefore are the focus of transmission-blocking drug and vaccine development. In order to better understand genes important to the sexual development process, the transcriptomes of high-purity stage I-V Plasmodium falciparum gametocytes were comprehensively profiled using a full-genome high-density oligonucleotide microarray. The interpretation of this transcriptional data was aided by applying a novel knowledge-based data-mining algorithm termed ontology-based pattern identification (OPI) using current information regarding known sexual stage genes as a guide. This analysis resulted in the identification of a sexual development cluster containing 246 genes, of which approximately 75% were hypothetical, exhibiting highly-correlated, gametocyte-specific expression patterns. Inspection of the upstream promoter regions of these 246 genes revealed putative cis-regulatory elements for sexual development transcriptional control mechanisms. Furthermore, OPI analysis was extended using current annotations provided by the Gene Ontology Consortium to identify 380 statistically significant clusters containing genes with expression patterns characteristic of various biological processes, cellular components, and molecular functions. Collectively, these results, available as part of a web-accessible OPI database (http://carrier.gnf.org/publications/Gametocyte), shed light on the components of molecular mechanisms underlying parasite sexual development and other areas of malarial parasite biology.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Plasmodium falciparum/genetics , Transcription, Genetic , Animals , Base Sequence , DNA, Protozoan/genetics , Female , Genome, Protozoan , Male , Multigene Family , Plasmodium falciparum/growth & development , Sequence Alignment , Sexual Maturation
17.
Mol Biochem Parasitol ; 142(2): 177-83, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15876462

ABSTRACT

The transcriptional repertoire of the in vivo liver stage of Plasmodium has remained largely unidentified and seemingly not amenable to traditional molecular analysis because of the small number of parasites and large number of uninfected hepatocytes. We have overcome this obstruction by utilizing laser capture microdissection to provide a high quality source of parasite mRNA for the construction of a liver stage cDNA library. Sequencing and annotation of this library demonstrated expression of 623 different Plasmodium yoelii genes during development in the hepatocyte. Of these genes, 25% appear to be unique to the liver stage. This is the first comprehensive analysis of in vivo gene expression undertaken for the liver stage of P. yoelii, and provides insights into the differential expression of P. yoelii genes during this critical stage of development.


Subject(s)
Gene Expression Regulation, Developmental , Liver/parasitology , Malaria/parasitology , Plasmodium yoelii/growth & development , Protozoan Proteins/metabolism , Animals , Expressed Sequence Tags , Gene Library , Hepatocytes/parasitology , Liver/cytology , Mice , Mice, Inbred BALB C , Plasmodium yoelii/genetics , Plasmodium yoelii/metabolism , Protozoan Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
18.
Science ; 307(5706): 82-6, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15637271

ABSTRACT

Plasmodium berghei and Plasmodium chabaudi are widely used model malaria species. Comparison of their genomes, integrated with proteomic and microarray data, with the genomes of Plasmodium falciparum and Plasmodium yoelii revealed a conserved core of 4500 Plasmodium genes in the central regions of the 14 chromosomes and highlighted genes evolving rapidly because of stage-specific selective pressures. Four strategies for gene expression are apparent during the parasites' life cycle: (i) housekeeping; (ii) host-related; (iii) strategy-specific related to invasion, asexual replication, and sexual development; and (iv) stage-specific. We observed posttranscriptional gene silencing through translational repression of messenger RNA during sexual development, and a 47-base 3' untranslated region motif is implicated in this process.


Subject(s)
Genome, Protozoan , Life Cycle Stages , Plasmodium/growth & development , Plasmodium/genetics , Proteome/analysis , 3' Untranslated Regions , Animals , Anopheles/parasitology , Computational Biology , Evolution, Molecular , Gene Expression Profiling , Gene Silencing , Genes, Protozoan , Malaria/parasitology , Oligonucleotide Array Sequence Analysis , Plasmodium/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Plasmodium chabaudi/genetics , Plasmodium chabaudi/growth & development , Plasmodium chabaudi/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Plasmodium yoelii/genetics , Plasmodium yoelii/growth & development , Plasmodium yoelii/metabolism , Proteomics , Protozoan Proteins/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Protozoan/genetics , RNA, Protozoan/metabolism , Selection, Genetic , Transcription, Genetic
19.
Genome Res ; 14(11): 2308-18, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15520293

ABSTRACT

To investigate the role of post-transcriptional controls in the regulation of protein expression for the malaria parasite, Plasmodium falciparum, we have compared mRNA transcript and protein abundance levels for seven different stages of the parasite life cycle. A moderately high positive relationship between mRNA and protein abundance was observed for these stages; the most common discrepancy was a delay between mRNA and protein accumulation. Potentially post-transcriptionally regulated genes are identified, and families of functionally related genes were observed to share similar patterns of mRNA and protein accumulation.


Subject(s)
Genes, Protozoan , Life Cycle Stages/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Protozoan Proteins/biosynthesis , Transcription, Genetic/genetics , Animals , Gene Expression Regulation/genetics , Plasmodium falciparum/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Protozoan/biosynthesis , RNA, Protozoan/genetics
20.
J Parasitol ; 90(5): 1062-71, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15562607

ABSTRACT

Using bioinformatic, proteomic, immunofluorescence, and genetic cross methods, we have functionally characterized a family of putative parasite ligands as potential mediators of cell-cell interactions. We name these proteins the Limulus clotting factor C, Coch-5b2, and Lgl1 (LCCL)-lectin adhesive-like protein (LAP) family. We demonstrate that this family is conserved amongst Plasmodium spp. It possesses a unique arrangement of adhesive protein domains normally associated with extracellular proteins. The proteins are expressed predominantly, though not exclusively, in the mosquito stages of the life cycle. We test the hypothesis that these proteins are surface proteins with 1 member of this gene family, lap1, and provide evidence that it is expressed on the surface of Plasmodium berghei sporozoites. Finally, through genetic crosses of wild-type Pblap1+ and transgenic Pblap1- parasites, we show that the null phenotype previously reported for sporozoite development in a Pblap1- mutant can be rescued within a heterokaryotic oocyst and that infectious Pblap1 sporozoites can be formed. The mutant is not rescued by coparasitization of mosquitoes with a mixture Pblap1+ and Pblap1- homokaryotic oocysts.


Subject(s)
Lectins/genetics , Multigene Family , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Amino Acid Sequence , Animals , Anopheles , Blotting, Western , Female , Fluorescent Antibody Technique, Indirect , Gene Expression , Lectins/chemistry , Mice , Microscopy, Phase-Contrast , Molecular Sequence Data , Plasmodium berghei/chemistry , Polymerase Chain Reaction , Protozoan Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...